Page 1 of 46 Diabetes

Clec16a, Nrdp1, and USP8 form a ubiquitindependent tripartite complex that regulates beta cell mitophagy

Gemma Pearson1; Biaoxin Chai1; Tracy Vozheiko1; Xueying Liu1; Malathi Kandarpa2; 3 1,4* Robert C. Piper ; and Scott A. Soleimanpour

1Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA. 2Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA. 3Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA 4Veterans Affairs Ann Arbor Health Care System, Ann Arbor, MI 48105, USA.

*Corresponding Author

Scott A. Soleimanpour, M.D., 1000 Wall Street, 5317 Brehm Tower, Ann Arbor, MI 48105. Phone: (734) 7630528 Fax: (734) 2328162 Email: [email protected]

Running Title: βcell mitophagy requires ubiquitin signaling

Word Count: 4448

Figures: 7

Diabetes Publish Ahead of Print, published online November 27, 2017 Diabetes Page 2 of 46

ABSTRACT

Mitophagy is a cellular quality control pathway, which is essential to eliminate unhealthy mitochondria. While mitophagy is critical to pancreatic βcell function, the posttranslational signals governing βcell mitochondrial turnover are unknown. Here we report that ubiquitination is essential for the assembly of a mitophagy regulatory complex, comprised of the E3 ligase Nrdp1, the deubiquitinase enzyme USP8, and Clec16a, a mediator of βcell mitophagy with unclear function. We discover that the diabetes Clec16a encodes an E3 ligase, which promotes nondegradative ubiquitin conjugates to direct its mitophagy effectors and stabilize the Clec16aNrdp1USP8 complex. Inhibition of the Clec16a pathway by the chemotherapeutic lenalidomide, a selective ubiquitin ligase inhibitor associated with new onset diabetes, impairs βcell mitophagy, oxygen consumption, and insulin secretion. Indeed, patients treated with lenalidomide develop compromised βcell function. Moreover, the βcell

Clec16aNrdp1USP8 mitophagy complex is destabilized and dysfunctional following lenalidomide treatment as well as following glucolipotoxic stress. Thus, the Clec16aNrdp1

USP8 complex relies on ubiquitin signals to promote mitophagy and maintain mitochondrial quality control necessary for optimal βcell function.

2 Page 3 of 46 Diabetes

Pancreatic βcells rely on mitochondrial bioenergetics to fuel glucosestimulated insulin

secretion (GSIS), which is essential for normal glucose homeostasis and is impaired in all

forms of diabetes (1). Beyond respiratory function, the mass of healthy mitochondria,

regulated by a balance of mitochondrial biogenesis and turnover, is vital to maintain energy

demands for proper stimulussecretion coupling. Mitochondrial autophagy (mitophagy) is a

quality control mechanism necessary for the selective elimination of dysfunctional

mitochondria to maintain optimal mitochondrial respiration in many cell types, including β

cells (2; 3). Despite the importance of mitophagy to βcell function (47), the molecular

signals dictating mitophagy and their significance to mitochondrial health remain largely

undefined.

Ubiquitination of substrates, catalyzed by the linkage of activated ubiquitin by

three distinct enzymes (E1, E2, E3), leads to diverse responses, including substrate

degradation by the ubiquitinproteasome system, protein trafficking, complex assembly, and

cellular signaling (8). Posttranslational modification of downstream mitophagy effectors,

including the E3 ligase Parkin, by ubiquitination is necessary for efficient mitochondrial

turnover (9); however, evidence of posttranslational control of proximal mitophagy

regulators has not been identified. The E3 ubiquitin ligase neuregulin receptor degradation

protein1 (Nrdp1) and the deubiquitination enzyme ubiquitin specific protease8 (USP8)

counterbalance ubiquitin dynamics to direct Parkin action or its proteasomal degradation (10;

11). Nrdp1 controls Parkinmediated mitophagy in βcells through its interaction with, and

regulation by, the diabetes gene C-type lectin domain family 16, member A (Clec16a) (4; 5);

however, the precise action of Clec16a in mitophagy remains poorly understood. While these

factors (Nrdp1, USP8, Clec16a) act upstream as sentinels to ensure mitophagy is utilized

appropriately, the role of ubiquitination as a control mechanism for their actions is unknown.

3 Diabetes Page 4 of 46

Here we discover that formation of a ubiquitindependent Clec16aNrdp1USP8 complex is necessary for control of mitophagy in βcells. We identify that inhibition of

Clec16amediated Nrdp1 ubiquitination by the chemotherapeutic agent lenalidomide impairs

Clec16amediated mitophagy and, ultimately, reduces βcell function. We demonstrate that

Clec16a is an E3 ubiquitin ligase, which ubiquitinates Nrdp1 to promote assembly of the

Clec16aNrdp1USP8 mitophagy complex. Lastly, we determine that mobilization and function of the βcell Clec16aNrdp1USP8 mitophagy complex is disrupted by glucolipotoxicity.

4 Page 5 of 46 Diabetes

RESEARCH DESIGN AND METHODS

Patient samples Samples were provided from deidentified nondiabetic multiple myeloma

patients by the University of Michigan Hematologic Malignancies Tissue Bank and in

compliance with the University of Michigan IRB. Fasting samples were collected prior to

chemotherapy and again 12 weeks later. Patients received a combination of Perifosine,

bortezomib, liposomal doxorubicin, Bexxar and/or carfilzomib in the presence/absence of

lenalidomide (25 mg PO daily for 28 days x 4 cycles). All patients received corticosteroids

(dexamethasone 40 mg PO daily x 12 doses/cycle). Cpeptide and glucose concentrations

were measured by chemiluminescence and the glucose oxidase method, respectively, by the

University of Michigan DRC Clinical Core. Homeostatic model assessment of βcell function

and insulin resistance were calculated by HOMA2 (12).

Animals- Animal studies were approved by the University of Michigan IACUC. Clec16aloxP

mice (5) were mated to MIPCreERT2 mice (13) to generate experimental groups.

Recombination was induced with 3 IP injections of 150µg/g BW tamoxifen (Cayman

Chemical) every other day over 5 days. 10week old C57BL/6 mice were injected daily with

50mg/kg of lenalidomide IP or vehicle control for 3weeks. Mice were left to recover for one

week before undergoing glucose tolerance testing. Animals were housed on a standard 12h

light/12h dark cycle with ad libitum access to food and water unless fasted prior to testing.

Antibodies A full listing of antibodies is provided in Supplementary Table 1.

Proximity ligation Studies were performed with a commercially available kit (Duolink;

Sigma) per manufacturers’ protocols utilizing Nrdp1 (Bethyl Labs) and USP8 (Sigma)

specific antisera. Counter staining to identify βcells was performed in dissociated human

5 Diabetes Page 6 of 46

islets utilizing PDX1 antisera (Santa Cruz). Proximity ligation events were quantified via

ImageJ similar to published approaches (14). Imaging was performed with an IX81 microscope (Olympus). Zstack images were captured using an ORCA Flash4 CMOS digital camera (Hamamatsu) and then subjected to deconvolution (CellSens; Olympus). Co localization analysis was performed as previously described (5).

Islet isolation and tissue culture - Primary dermal fibroblasts from WT and Ubc

CreERT;Clec16alox/lox mice were treated ex vivo with tamoxifen as previously described (5).

Stably expressing FlagClec16a NIH3T3 cells, and Min6 cells (a gift from Dr. Doris Stoffers) were maintained as previously described (5). Mouse islets were isolated and cultured as previously described (15). Cell treatments included dimethylsulfoxide (DMSO; Fisher), lenalidomide (Ark Pharm), chloroquine (Santa Cruz Biotechnology), FCCP (Sigma), bafilomycin A1, MG132, and cycloheximide (all from EMD Millipore). Palmitate/BSA couplings were generated as previously described (16).

Human islets– Studies performed on human islets procured from the Integrated Islet

Distribution Program (IIDP) were approved by the University of Michigan IRB. Donor information is provided in Supplementary Table 2. Human islets were cultured in PIM(S)

(Prodo Laboratories) supplemented with 10% FBS, 1% antibiotic/antimycotic (Life

Technologies), and 1% PIM(G) (Prodo Laboratories). Following culture and treatments, islets were dissociated into single cells using trypsin, cytocentrifuged to glass slides, and prepared for immunostaining.

Transfections Transfection of HEK293T, HeLa, and NIH3T3 cells (Lipofectamine 2000;

Life Technologies) and Min6 cells (Amaxa Nucleofector) was performed as previously

6 Page 7 of 46 Diabetes

described (5). A detailed list of plasmids utilized or generated for this study is found in online

supplementary material.

Protein expression/purification Recombinant Clec16a and Nrdp1 was expressed in

NiCo21 (DE3) strain E.coli (NEB) following culture in autoinduction media as described

(17). GSTtagged and 6xHistagged were purified utilizing glutathione matrix or

nickelcharged resin (HiCap and NiNTA agarose; Qiagen) per manufacturers’ protocols. In

vitro transcribed/translated Clec16a and Nrdp1 were expressed in T7driven mammalian

expression systems (Promega) per manufacturers’ protocols. Clec16a and Nrdp1 protein

expression/purification was confirmed by silver staining (BioRad) and/or Western blotting.

Ubiquitination assays Detection of protein ubiquitination in vivo was performed in cell

lines similar to previously described approaches (18). Detection of protein ubiquitination in

vitro was performed following incubation of 1µM GSTClec16aFlag or Clec16a6xHisFlag

with a commercially available kit (E2 Select; BioTechne). Ubiquitin conjugation assays

were performed with 1µM GSTClec16aFlag/Clec16a6xHisFlag, 2.5µM GSTNrdp1

CSHQ, 50nM E1, 5µM Ube2d3/Ube2c, 50µM HAubiquitin, MgATP, and conjugation

reaction buffers (BioTechne). Conjugation assays were performed in solution for 60 minutes

at 37°C. Ubiquitination and protein interaction assays from in vitro transcribed/translated

FlagClec16a and HANrdp1 were incubated in 40mM TrisHCl (pH 7.6), 5mM MgCl2,

2mM DTT with/without the addition of 75µM PYR41 (EMD Millipore) for 3 hours at 37°C

similar to previously published approaches (19).

7 Diabetes Page 8 of 46

Glucose/insulin measurements, respirometry, gene expression, immunoprecipitation, immunofluorescence, and Western blotting assays All assays were performed as previously described (5).

Statistics Data are presented as mean ± SEM, and error bars denote SEM. Statistical comparisons were performed using one or twotailed Student’s ttest or twoway ANOVA as appropriate (Prism GraphPad). A P value < 0.05 was considered significant.

8 Page 9 of 46 Diabetes

RESULTS

Inhibition of Nrdp1 ubiquitination impairs human βcell function and mitophagy.

We hypothesized that ubiquitination of upstream mitophagy regulators regulates βcell

mitochondrial quality control to preserve the wellfunctioning mitochondria necessary for

proper stimulussecretion coupling and GSIS. To this end, we assayed βcell function in

multiple myeloma patients who received lenalidomide, a chemotherapeutic agent and

selective ubiquitin ligase inhibitor (20), which, along with its parent compound thalidomide,

is associated with newonset glucose intolerance (21; 22). Lenalidomide has also been shown

to impair ubiquitination of the E3 ligase Nrdp1 (23), an essential regulator of βcell

mitophagy (4; 5). To evaluate the effect of lenalidomide in vivo, we estimated βcell function

by HOMAβ in patients treated with or without lenalidomide for 12 weeks (Fig 1A and

Supplementary Table 3). Although all patients received glucocorticoids as part of their

treatment regimen (see methods), none had prior documented diabetes or glucose intolerance.

Interestingly, we observed decreased HOMAβ (Fig 1A) in lenalidomidetreated patients.

Impaired βcell function was not secondary to advanced age or increased insulin resistance,

as lenalidomidetreated patients were younger and had no differences in HOMAIR

compared to controls (Supplementary Table 3 and data not shown). We also observed that

shortterm intraperitoneal lenalidomide treatment (3 weeks) induced modest glucose

intolerance in mice (Fig S1AB); however, the milder effects of lenalidomide in mice could

be attributed to the rapid clearance of intraperitoneal lenalidomide (versus oral) as well as a

shorter duration of exposure (24). We also confirmed that lenalidomide impaired

ubiquitination of Nrdp1 in Min6 βcells (Fig S1C). Further, lenalidomide significantly

reduced both GSIS and oxygen consumption in isolated islets (Fig 1BC).

Given the importance of Nrdp1 to mitophagy and maintenance of insulin secretion,

we hypothesized that lenalidomide would lead to dysfunctional mitophagy in βcells. Indeed,

9 Diabetes Page 10 of 46

the βcells of lenalidomidetreated mice developed an accumulation of autophagosomes containing mitochondria, as shown by increased colocalization of the autophagosome marker LC3 with the mitochondrial protein succinate dehydrogenase A (SDHA; Fig 1DE).

Additionally, lenalidomidetreated islets developed an increase in the total number of LC3+ puncta (Fig 1E) without overt changes in LC3I/II protein levels (Fig 1F), suggesting lenalidomide disrupts selective rather than bulk autophagy. As a complementary approach, we investigated the effects of lenalidomide on mitophagic flux by assessing the co localization of mitochondria with autophagosomes using fluorescentlabeled markers

(mitochondria mitodsRed; autophagosomes – LC3GFP) in Min6 βcells, at baseline and following mitochondrial damage by the uncoupler FCCP (5). Concordantly, lenalidomide treatment led to increased colocalization of LC3GFP and mitoDsRed (Fig 1GH). It is unlikely that lenalidomide enhances mitophagic flux however, as the increased co localization of mitodsRed and LC3GFP at baseline is not maintained after FCCP treatment

(Fig 1GH), suggesting that dysfunctional mitochondria are not efficiently removed. Taken together, we observe that lenalidomide impairs glucose homeostasis, GSIS, mitochondrial respiration, and Nrdp1 ubiquitination, and our results implicate the importance of Nrdp1 ubiquitination to maintain mitophagy and βcell function.

Clec16a promotes ubiquitination of Nrdp1.

Protein ubiquitination leads to several posttranslational effects, including targeting for proteasomal degradation, trafficking, or the activation of downstream signaling cascades.

Our observation of a lenalidomidesensitive effect on Nrdp1 ubiquitination and βcell mitophagy led us to assess the role of Nrdp1 interaction partners that have downstream consequences on mitophagy. Nrdp1 interacts with two upstream regulators of mitophagy:

Clec16a and the deubiquitinase (DUB) enzyme USP8 (5; 25). Specifically, Clec16a prevents

10 Page 11 of 46 Diabetes

the proteasomal degradation of Nrdp1 to control downstream events in βcell mitophagy (5);

therefore, we investigated the role of Clec16a on Nrdp1 stability and ubiquitination. Clec16a

deficiency decreased Nrdp1 levels and protein stability (Figs S2A, C) while Clec16a

overexpression increased Nrdp1 protein accumulation, consistent with previous reports in β

cells (4). Surprisingly, we observed that Clec16a overexpression resulted in enhanced Nrdp1

ubiquitination, while Clec16aKO primary fibroblasts exhibited reduced Nrdp1 ubiquitination

(Figs 2AC, Fig S2B).

Clec16amediated enhancement of Nrdp1 ubiquitination and stability is in contrast to

the effects of USP8, a DUB enzyme which stabilizes Nrdp1 by opposing its auto

ubiquitination and preventing proteasomal degradation (Fig 2A and (25)). We tested the

importance of USP8 to Clec16amediated Nrdp1 stability by both gain and loss of function

approaches. We reproduced published findings of enhanced Nrdp1 levels following USP8

overexpression (Fig S2D) (25). We observed no changes in basal Nrdp1 levels with USP8

shRNA (Fig S2E), consistent with previous studies (26), possibly suggesting other factors

stabilize Nrdp1 following USP8 loss of function. Clec16a increased Nrdp1 levels

independent of USP8 levels (Fig S2DE), suggesting that USP8 and Clec16a act via distinct,

yet complementary means to promote Nrdp1 protein accumulation. While Clec16a is an

endolysosomal protein, it also appears to promote Nrdp1 levels independent of lysosomal

degradation (Fig S2F). To investigate whether Clec16a promotes Nrdp1 ubiquitination by

enhancing the autoubiquitination activity of Nrdp1, we generated an Nrdp1 RINGdomain

mutant (C34S H36Q, also known as Nrdp1 CSHQ) lacking E3 ligase activity (27). In the

presence of Clec16a, both Nrdp1WT and Nrdp1CSHQ exhibited enhanced ubiquitination

(Fig 2D), suggesting that Clec16a promotes Nrdp1 ubiquitination independent of Nrdp1 self

ubiquitination.

11 Diabetes Page 12 of 46

The similar effects of Clec16a loss of function and lenalidomide treatment on Nrdp1 ubiquitination led us to hypothesize that lenalidomide may act through Clec16a to impact β cell function. Indeed, lenalidomide abrogated Clec16amediated Nrdp1 ubiquitination (Fig

2E). To investigate how Clec16amediated Nrdp1 ubiquitination affects βcell function, we studied tamoxifeninducible and βcellspecific Clec16a knockout mice (MIPCreERT2;

Clec16aloxP/loxP, hereafter known as βClec16aKO). βClec16aKO mice exhibited reduced

Clec16a mRNA levels and impaired glucose tolerance similar to previous reports (Fig S3A

C; (5)). As expected, lenalidomide treatment or Clec16a deficiency resulted in decreased

GSIS compared to vehicletreated MIPCreERT2 controls (Fig 2F). Importantly, we did not observe any additive impairment of lenalidomide on βClec16aKO islets (Fig 2F), suggesting that lenalidomide acts through the Clec16a pathway to regulate Nrdp1 ubiquitination and β cell function. Notably, lenalidomide treatment did not affect Clec16a mRNA and protein levels (Fig 2E and S3D). These results suggest that lenalidomide inhibits Clec16a ubiquitination of Nrdp1, and that this activity is essential for downstream maintenance of maximal glucosestimulated insulin release from βcells.

Clec16a is an E3 ubiquitin ligase.

Despite the connection between Clec16a and mitophagy/autophagy (5; 28; 29), the direct molecular/enzymatic function for Clec16a has remained elusive. We questioned whether

Clec16a possesses intrinsic ubiquitin ligase activity and investigated its capacity to induce ubiquitination in vitro. A vital characteristic of E3 ubiquitin ligases is their ability to auto ubiquitinate (30), and thus we screened 26 independent E2 conjugation enzymes for their ability to participate in Clec16a autoubiquitination. Recombinant Clec16a protein was incubated with each E2 enzyme, and evaluated for the presence of an increased molecular weight shift by SDSPAGE, which would be suggestive of ubiquitinated Clec16a.

12 Page 13 of 46 Diabetes

Interestingly, we observed that both Ube2C and Ube2D3 were able to promote high MW

Clec16a protein (Fig 3A). Concordantly, we observed that recombinant Clec16a (Fig 3BC)

was sufficient to promote autoubiquitination in an ATP, Ube2C/Ube2D3, and ubiquitin

dependent manner. Moreover, addition of Clec16a to an autoubiquitinationdeficient Nrdp1

mutant (Nrdp1CSHQ) led to enhanced Nrdp1 ubiquitination, suggesting that Clec16a

directly ubiquitinates Nrdp1 (Fig 3D).

Ubiquitination leads to diverse cellular effects, dependent in part upon the linkages in

the ubiquitin chain. Linkages can occur on one of seven distinct ubiquitin lysine residues and

proteasomal degradation generally only follows K48linked ubiquitination (8). Furthermore,

K48independent ubiquitination plays crucial signaling roles in mitophagy (10; 31; 32).

Given that Clec16a promotes Nrdp1 protein accumulation, we postulated that Clec16a might

link nondegradative ubiquitin moieties to Nrdp1. Clec16a potentiated wildtype (WT)

ubiquitin linkages on Nrdp1, which was lost following the addition of a ubiquitin mutant

rendered incapable of ubiquitin laddering by mutagenesis of all 7 lysine residues to arginine

(KO; Fig 3E). Furthermore, Clec16a potentiated Nrdp1 ubiquitination in the presence of a

ubiquitin mutant incapable of K48degradative linkages (K48R), but not in the presence of

mutants capable of K48 and K63 linkages only (K48 and K63; Fig 3E). This suggests that

Clec16a relies upon K48 and K63independent nondegradative ubiquitin linkages to post

translationally regulate Nrdp1. Taken altogether, Clec16a is an E3ligase, which directly

ubiquitinates its mitophagy partner and substrate Nrdp1 with nondegradative conjugates.

Clec16a, USP8, and Nrdp1 interact in a ubiquitindependent complex.

The convergence of Clec16a and USP8 in the regulation of Nrdp1 and control of mitophagy,

led us to question whether Clec16a, Nrdp1, and USP8 occupy the same subcellular

compartment to participate in a tripartite complex. Clec16a is an endolysosomal protein and

13 Diabetes Page 14 of 46

may colocalize with Nrdp1 and USP8, which have been previously observed within the endosomal compartment (33). Indeed, we observed that Clec16a as well as Nrdp1 and USP8 were isolated within the membrane protein fraction (which contains endolysosomal proteins) following differential centrifugation (Fig 4A). Furthermore, we observed Clec16a, Nrdp1, and USP8 colocalization by immunostaining, (Fig 4B), indicating they do indeed occupy a common compartment.

To determine the role of ubiquitination on complex assembly, we studied the interaction of in vitro transcribed/translated Clec16a and Nrdp1 expressed in reticulocyte lysates and verified their interaction in this system (Fig 4C). We next coincubated in vitro derived Clec16a and Nrdp1 with the E1 initiation inhibitor PYR41 to test the role of ubiquitination independent of its cytotoxic effects observed in vivo (19). PYR41 treatment reduced the interaction between Clec16a and Nrdp1 (Fig 4D), implicating that ubiquitination maintains the Clec16aNrdp1 complex. We next determined the role of ubiquitination and

Clec16a in stabilization of Nrdp1USP8 binding utilizing wildtype and mutant ubiquitin.

While Clec16a enhanced Nrdp1USP8 complex assembly, addition of the ubiquitin KO mutant reduced Nrdp1 levels as well as Nrdp1 interaction with both Clec16a and USP8 (Fig

4E). To clarify if impaired ubiquitination affected the Clec16aNrdp1USP8 complex due to reduced levels of constituents or by impaired complex assembly, we also doubled the amount of FlagClec16a plasmid in the presence of KO ubiquitin, resulting in increased levels of

Clec16a, USP8 and Nrdp1 (Fig 4E, lane ++). While levels of constituent proteins were rescued, interactions with Nrdp1 were only partially restored with Clec16a, yet not with

USP8. Collectively, these findings suggest that functional ubiquitination is necessary for maintaining levels of the constituents of the Clec16aNrdp1USP8 complex as well as providing vital signals for complex assembly.

14 Page 15 of 46 Diabetes

USP8 shares functional overlap with Clec16a/Nrdp1 in the regulation of mitophagic

flux, coinciding in their opposing regulation of the E3 ligase Parkin, a critical effector in

mitophagy (34). While Clec16a and Nrdp1 limit excessive Parkin activity to ensure healthy

mitochondria are not degraded (5), USP8 promotes Parkin translocation to the mitochondrial

outer membrane by removal of K6linked ubiquitin conjugates (10). Therefore, we

investigated whether Clec16a influences the K6linked ubiquitination state of Parkin.

Utilizing a mutant ubiquitin only capable of K6linked conjugates, we observed the expected

reduction of K6linked Parkin ubiquitination by USP8 (Fig 4F). Conversely, Clec16a

promotes K6linked Parkin ubiquitination independent of USP8 (Fig 4F) indicating that

Clec16a opposes USP8mediated K6deubiquitination of Parkin. Consequently, we conclude

that Clec16aNrdp1USP8 assemble in a tripartite ubiquitindependent mitophagy complex to

promote maintenance of Parkin.

Nrdp1 ubiquitination is pivotal for complex formation and βcell function

Given the importance of ubiquitination to Clec16aNrdp1USP8 complex assembly, we

hypothesized that specific ubiquitination of Nrdp1 may influence formation of the Clec16a

Nrdp1USP8 complex. To investigate how Nrdp1 ubiquitination could influence Clec16a

mediated processes, we generated an Nrdp1 mutant (Nrdp1 KR), in which all 11 lysine

residues were mutated to arginine, thereby rendering it incapable of ubiquitination. As

expected, ubiquitin laddering was not observed on the Nrdp1 KR mutant despite Clec16a

overexpression (Fig. 5A). Not surprisingly, overall levels of the Nrdp1 KR mutant protein

were higher that WT Nrdp1, potentially due to a loss of degradative Nrdp1 auto

ubiquitination (Fig. 5A; (25)). Furthermore, Clec16a was incapable of increasing Nrdp1

levels in the presence of the KR mutant, reinforcing the importance of Clec16amediated

Nrdp1 ubiquitination to enhance Nrdp1 stability (Fig. 5A). We next assessed the role of

15 Diabetes Page 16 of 46

Nrdp1 ubiquitination on formation of the Clec16aNrdp1USP8 complex. Interestingly,

Clec16a interacts with endogenous USP8 and this interaction is maintained in the presence or absence of Nrdp1 ubiquitin ligase activity (Fig. 5B). However, Clec16aUSP8 binding as well as USP8 levels are reduced following overexpression of the Nrdp1 KR mutant (Fig.

5B). These results suggest, again, that Clec16a drives assembly of the Clec16aNrdp1USP8 complex by providing essential ubiquitin signals and stabilizing its component proteins.

To determine whether the Clec16aNrdp1USP8 axis regulates βcell function, we assessed the role of Clec16amediated ubiquitination on GSIS in Min6 βcells. We found that shRNAmediated reduction of Clec16a decreased GSIS (Figure S3EF). Clec16a shRNA treatment also led to reduced WTNrdp1 levels, whereas Clec16a does not modulate levels of the ubiquitindeficient Nrdp1 KR mutant (Fig. 5C). Overexpression of WTNrdp1, but not

Nrdp1 KR, restored GSIS in shClec16atreated Min6 βcells (Fig. 5D), suggesting that

Clec16amediated ubiquitination of Nrdp1 is necessary for optimal βcell function. To assess the role of Clec16amediated ubiquitination on formation of the βcell mitophagy complex, we utilized a proximity ligation assay (PLA) to visualize Nrdp1USP8 association in situ following lenalidomide treatment (35). Lenalidomide impaired Nrdp1USP8 interaction in

Min6 βcells (Fig. 5E) suggesting Clec16amediated ubiquitination maintains the Clec16a

Nrdp1USP8 complex as well as βcell function.

The Clec16aNrdp1USP8 complex is perturbed by glucolipotoxicity

Defects in βcell mitochondrial structure and function have been observed under conditions of diabetogenic stimuli, notably glucolipotoxicity (36; 37). To determine if the combination of reduced Clec16a expression and/or glucolipotoxicity impairs the upstream mitophagy complex, we assessed Nrdp1USP8 interaction by PLA. As expected, Clec16a specific shRNA reduced Nrdp1USP8 binding in situ (Fig. 6A). Nrdp1USP8 binding was

16 Page 17 of 46 Diabetes

also decreased following palmitate treatment, indicating that glucolipotoxicity destabilizes

upstream mitophagy regulators (Fig 6A). However, palmitate treatment did not additively

reduce Nrdp1USP8 binding in shClec16atreated Min6 βcells (Fig 6A), suggesting that

palmitate acts to destabilize the Nrdp1USP8 complex through effects on the Clec16a

pathway. We next investigated the impact of glucolipotoxicity on the Clec16NrdpUSP8

complex in human islets. Similarly, glucolipotoxicity decreased Nrdp1USP8 interaction in

primary human βcells (Fig 6B). Given our findings suggesting that both Nrdp1 stability and

ubiquitination regulates formation of the Clec16aNrdp1USP8 complex, we next measured

Nrdp1 levels following palmitate treatment in primary islets. Indeed, we observed a

significant reduction in Nrdp1 levels during glucolipotoxic conditions (Fig 6C). Furthermore,

we observed that both Nrdp1 stability and ubiquitination was reduced in Min6 βcells

following palmitate treatment (Fig 6D), thus demonstrating effects consistent with impact on

the Clec16apathway to maintain the upstream mitophagy complex.

To determine if glucolipotoxicity affects Clec16a to impact formation of the Clec16a

Nrdp1USP8 complex, we overexpressed Clec16a in palmitatetreated Min6 βcells.

Importantly, Clec16a overexpression in palmitatetreated cells raised Nrdp1 levels, Nrdp1

ubiquitination, and Nrdp1USP8 binding back to baseline levels (BSA alone; Fig. 6DE).

While palmitatetreated Clec16a overexpressing cells did not rescue the above parameters

completely to the same degree as Clec16a overexpression alone, it is noteworthy that

palmitate treatment itself reduced levels of Clec16a (Fig. 6DE), suggestive of an effect of

palmitate on Clec16a.

Beyond mitochondrial functional defects, chronic glucolipotoxicity can ultimately

promote βcell apoptosis (1; 36). To determine the importance of maintenance of the

Clec16aNrdp1USP8 complex to βcell survival during glucolipotoxicity, we assessed βcell

apoptosis by cleaved caspase3 levels following Clec16a overexpression. As expected,

17 Diabetes Page 18 of 46

palmitate significantly induced βcell apoptosis in Min6 βcells (Fig. 6F). Interestingly,

Clec16a overexpression reduced cleaved caspase3 levels following palmitate treatment (Fig.

6F), suggesting that restoration of the Clec16apathway partially ameliorates the negative effects of glucolipotoxicity on βcell survival. Taken altogether, these results indicate that glucolipotoxic insults destabilize the ubiquitindependent Clec16Nrdp1USP8 mitophagy complex, which could ultimately impact the ability of βcells to withstand noxious stimuli.

DISCUSSION

Here we describe that ubiquitination is essential for the function and assembly of upstream mitophagy regulators in their maintenance of βcell function. Pharmacologic impairment of

Clec16amediated ubiquitination by the chemotherapeutic agent lenalidomide impairs mitophagy and insulin release. We identify that the diabetes gene Clec16a encodes an E3 ligase that promotes Nrdp1 ubiquitination. Furthermore, Clec16amediated Nrdp1 ubiquitination is essential for assembly of the Clec16aNrdp1USP8 complex and preservation of βcell function (Fig 7). We observe that glucolipotoxicity perturbs the assembly and function of this complex in βcells. Thus, Clec16aNrdp1USP8 function in a ubiquitindependent protein complex to regulate βcell mitophagy.

Posttranslational control of Parkin by ubiquitination and phosphorylation is necessary for the execution of mitophagy (38). However, the direct convergence of Clec16a,

Nrdp1, and USP8, and the dependence on ubiquitination for upstream control of mitophagy have not been previously appreciated. The tripartite Clec16aNrdp1USP8 complex we identify coordinates mitophagy in part through their integrated actions on Parkin and cohesively harmonizes the effects of these mitophagy regulators (5; 10; 25). Several DUB enzymes, including USP8, regulate Parkin action during mitophagy (3941). While Parkin autoinhibition and K6ubiquitination are relieved by USP8 to promote its mitochondrial

18 Page 19 of 46 Diabetes

localization, it is unclear how Clec16a opposes USP8mediated deubiquitination of Parkin.

Nrdp1 binds and opposes USP8 action on cytokine receptor signaling (42), and Clec16a

could function through Nrdp1 to counter USP8 effects on Parkin. Clec16a may also directly

(or indirectly through other E3 ligases) promote K6linked conjugates on Parkin. While the

dynamics of the Clec16aNrdp1USP8 complex are still a mystery, it is clear that Clec16a

mediated ubiquitination orchestrates mitophagic flux in βcells.

Our discovery of Clec16a as an E3 ligase, whose function is inhibited by

lenalidomide, expands our insight into its functional role in mitophagy. Initial prediction of

Clec16a function was challenging due to a lack of recognizable conserved motifs, including

the misidentified predicted Clectin domain, suggesting Clec16a is a misnomer (5; 43). While

it is clear that Clec16a has E3 ligase activity, we could not identify a consensus domain

consistent with known ubiquitin ligases. Indeed, future studies of Clec16a domains coupled

to structural prediction of lenalidomideClec16a binding sites may reveal a potential ubiquitin

ligase domain. Additionally, lenalidomide inhibits other E3ligases, notably Cereblon (20).

Cereblon function in βcells is unknown, therefore additional study is needed to determine

the contribution of Cereblon and/or Clec16a inhibition to βcell dysfunction caused by

lenalidomide in humans.

Clec16a ubiquitinates its principal partner Nrdp1, leading to ubiquitin laddering of

unknown composition, which could occur at one or several lysine residues. Clec16amediated

Nrdp1 ubiquitin conjugates may also be modified by the cooperative actions of USP8 to

favor nondegradative linkages while removing K48 linkages. Alternatively, there may be

Clec16aindependent effects to Nrdp1 activity that regulate βcell function. Notably, the ER

structural protein Reticulon 4A (Rtn4a or NogoA) as well as retinoic acid derivatives

modulate Nrdp1 activity (44; 45) in nonβcells, and they do not intersect with the Clec16a

pathway in our previous interactome studies (5). Rtn4a has also been implicated in the

19 Diabetes Page 20 of 46

regulation of GSIS (46). Therefore, elucidating Clec16a/USP8dependent and independent effects on Nrdp1 function will provide insight into shared and unique pathways that could be evaluated for their contribution to βcell function.

Single nucleotide polymorphisms (SNPs) within the CLEC16A on

16 are associated with diabetes as well as several human diseases of both autoimmune and nonautoimmune origin (47; 48). We previously described that a diabetogenic SNP within the

CLEC16A locus correlated with impaired βcell function, diminished glycemic control, and functioned as an expression quantitative trait locus for human islet CLEC16A levels, while not affecting levels of other nearby ((5) and data not shown). While a diabetogenic

CLEC16A SNP does correlate with abnormal βcell function, we can only speculate that these

SNPs directly lead to abnormalities within the CLEC16Amitophagy pathway in human β cells. Clec16a is also a ubiquitously expressed gene with functions in other cell types, including immune cells and neurons (5; 28; 49). The effect of diseaserelated CLEC16A

SNPs to influence CLEC16A expression or the expression of nearby related genes in immune cells still remains to be controversial (48). While Clec16a and Nrdp1 both play vital roles within immune cells (28; 50), the function of CLEC16A SNPs (on mitophagy or other processes) within other human cell types requires future investigation.

These studies clarify a role for ubiquitindependent mitophagy complex assembly in the maintenance of βcell function. Mitochondrial competence is essential for βcell function, and deficient mitochondrial quality control could exacerbate βcell dysfunction by diabetogenic insults. Previous observations of defective mitochondrial integrity following glucolipotoxicity (7; 36), coupled to our findings here of glucolipotoxicitymediated destabilization of the Clec16aNrdp1USP8 complex could inspire future in vivo studies to evaluate the importance of dysfunctional mitophagy to βcell failure in diabetes. Additional examination will be required to determine if nutrient excess, insulin resistance, and/or

20 Page 21 of 46 Diabetes

inflammation (among other diabetogenic stressors) interfere with the mitophagy machinery,

but may prove to illuminate a more profound understanding of diabetes pathogenesis.

21 Diabetes Page 22 of 46

ACKNOWLEDGEMENTS

We acknowledge Dr. D. Giacherio and J. Whalen of the Chemistry Laboratory Core of the

University of Michigan DRC (P30DK020572) for assistance with human hormone studies.

We thank Drs. K. Claiborn, J. Spaeth, P. Arvan, L. Satin, L. Qi, M. Myers, R. Stein, and members of the Soleimanpour laboratory for helpful advice. G.P. conceived of, designed and performed experiments, interpreted results, drafted and reviewed the manuscript. B.C., T.V.,

X.L. designed and performed experiments and interpreted results. M.K. provided human samples from the UM Hematologic Malignancies Tissue Bank. R.C.P. designed ubiquitination studies, interpreted results and reviewed the manuscript. S.A.S. conceived of and designed the studies, interpreted results, edited and reviewed the manuscript. S.A.S. is the guarantor of this work and takes responsibility for the contents of this article. All authors declare that they have no conflicts of interest. We acknowledge funding support from the

JDRF (CDA2016189, SRA2018539), the NIH (K08DK089117, R03DK106304, and

R01DK108921), the Central Society for Clinical and Translational Research, the Brehm family, and the Anthony family. The JDRF Career Development Award to S.A.S. is partly supported by the Danish Diabetes Academy supported by the Novo Nordisk Foundation.

22 Page 23 of 46 Diabetes

REFERENCES

1. Supale S, Li N, Brun T, Maechler P: Mitochondrial dysfunction in pancreatic β cells. Trends in Endocrinology & Metabolism 2012;23:477487 2. Jin SM, Youle RJ: PINK1 and Parkinmediated mitophagy at a glance. Journal of Cell Science 2012;125:795799 3. Kaufman BA, Li C, Soleimanpour SA: Mitochondrial regulation of βcell function: Maintaining the momentum for insulin release. Molecular Aspects of Medicine 2015; 4. Soleimanpour SA, Ferrari AM, Raum JC, Groff DN, Yang J, Kaufman BA, Stoffers DA: Diabetes Susceptibility Genes Pdx1 and Clec16a Function in a Pathway Regulating Mitophagy in βCells. Diabetes 2015;64:34753484 5. Soleimanpour Scott A, Gupta A, Bakay M, Ferrari Alana M, Groff David N, Fadista J, Spruce Lynn A, Kushner Jake A, Groop L, Seeholzer Steven H, Kaufman Brett A, Hakonarson H, Stoffers Doris A: The Diabetes Susceptibility Gene Clec16a Regulates Mitophagy. Cell 2014;157:15771590 6. Hoshino A, Ariyoshi M, Okawa Y, Kaimoto S, Uchihashi M, Fukai K, IwaiKanai E, Ikeda K, Ueyama T, Ogata T, Matoba S: Inhibition of p53 preserves Parkinmediated mitophagy and pancreatic βcell function in diabetes. Proceedings of the National Academy of Sciences 2014;111:31163121 7. Twig G, Elorza A, Molina AJA, Mohamed H, Wikstrom JD, Walzer G, Stiles L, Haigh SE, Katz S, Las G, Alroy J, Wu M, Py BF, Yuan J, Deeney JT, Corkey BE, Shirihai OS: Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. The EMBO Journal 2008;27:433446 8. Wong E, Cuervo AM: Integration of Clearance Mechanisms: The Proteasome and Autophagy. Cold Spring Harbor Perspectives in Biology 2010;2 9. Durcan TM, Fon EA: The three ‘P’s of mitophagy: PARKIN, PINK1, and post translational modifications. Genes Dev 2015;29:989999 10. Durcan TM, Tang MY, Pérusse JR, Dashti EA, Aguileta MA, McLelland GL, Gros P, Shaler TA, Faubert D, Coulombe B, Fon EA: USP8 regulates mitophagy by removing K6 linked ubiquitin conjugates from parkin. The EMBO Journal 2014;33:24732491 11. Zhong L, Tan Y, Zhou A, Yu Q, Zhou J: RING finger ubiquitinprotein isopeptide ligase Nrdp1/FLRF regulates parkin stability and activity. Journal of Biological Chemistry 2005;280:94259430 12. Wallace TM, Levy JC, Matthews DR: Use and Abuse of HOMA Modeling. Diabetes Care 2004;27:14871495 13. Tamarina NA, Roe MW, Philipson LH: Characterization of mice expressing Ins1 gene promoter driven CreERT recombinase for conditional gene deletion in pancreatic βcells. Islets 2014;6:e27685 14. McKenna B, Guo M, Reynolds A, Hara M, Stein R: Dynamic Recruitment of Functionally Distinct Swi/Snf Chromatin Remodeling Complexes Modulates Pdx1 Activity in Islet β Cells. Cell Reports 2015;10:20322042 15. Pearson GL, Mellett N, Chu KY, Cantley J, Davenport A, Bourbon P, Cosner CC, Helquist P, Meikle PJ, Biden TJ: Lysosomal acid lipase and lipophagy are constitutive negative regulators of glucosestimulated insulin secretion from pancreatic beta cells. Diabetologia 2014;57:129139 16. Busch AK, Cordery D, Denyer GS, Biden TJ: Expression Profiling of Palmitate and OleateRegulated Genes Provides Novel Insights Into the Effects of Chronic Lipid Exposure on Pancreatic bCell Function. Diabetes 2002;51:977987

23 Diabetes Page 24 of 46

17. Studier FW: Stable Expression Clones and AutoInduction for Protein Production in E. coli. In Structural Genomics: General Applications Chen YW, Ed. Totowa, NJ, Humana Press, 2014, p. 1732 18. Choo YS, Zhang Z: Detection of Protein Ubiquitination. Journal of Visualized Experiments : JoVE 2009:1293 19. Yang Y, Kitagaki J, Dai RM, Tsai YC, Lorick KL, Ludwig RL, Pierre SA, Jensen JP, Davydov IV, Oberoi P, Li CCH, Kenten JH, Beutler JA, Vousden KH, Weissman AM: Inhibitors of UbiquitinActivating Enzyme (E1), a New Class of Potential Cancer Therapeutics. Cancer Research 2007;67:94729481 20. Krönke J, Udeshi ND, Narla A, Grauman P, Hurst SN, McConkey M, Svinkina T, Heckl D, Comer E, Li X, Ciarlo C, Hartman E, Munshi N, Schenone M, Schreiber SL, Carr SA, Ebert BL: Lenalidomide Causes Selective Degradation of IKZF1 and IKZF3 in Multiple Myeloma Cells. Science 2014;343:301305 21. Issa ZA, Zantout MS, Azar ST: Multiple Myeloma and Diabetes. ISRN Endocrinology 2011;2011:6 22. Iqbal N, Zayed M, Boden G: Thalidomide impairs insulin action on glucose uptake and glycogen synthesis in patients with type 2 diabetes. Diabetes Care 2000;23:11721176 23. Basiorka AA, McGraw KL, De Ceuninck L, Griner LN, Zhang L, Clark JA, Caceres G, Sokol L, Komrokji RS, Reuther GW, Wei S, Tavernier J, List AF: Lenalidomide Stabilizes the Erythropoietin Receptor by Inhibiting the E3 Ubiquitin Ligase RNF41. Cancer Res 2016;76:35313540 24. Rozewski DM, Herman SEM, Towns WH, Mahoney E, Stefanovski MR, Shin JD, Yang X, Gao Y, Li X, Jarjoura D, Byrd JC, Johnson AJ, Phelps MA: Pharmacokinetics and Tissue Disposition of Lenalidomide in Mice. The AAPS Journal 2012;14:872882 25. Wu X, Yen L, Irwin L, Sweeney C, Carraway KL: Stabilization of the E3 Ubiquitin Ligase Nrdp1 by the Deubiquitinating Enzyme USP8. Mol Cell Biol 2004;24:77487757 26. Cao Z, Wu X, Yen L, Sweeney C, Carraway KL: NeuregulinInduced ErbB3 Downregulation Is Mediated by a Protein Stability Cascade Involving the E3 Ubiquitin Ligase Nrdp1. Mol Cell Biol 2007;27:21802188 27. Qiu XB, Goldberg AL: Nrdp1/FLRF is a ubiquitin ligase promoting ubiquitination and degradation of the epidermal growth factor receptor family member, ErbB3. Proceedings of the National Academy of Sciences 2002;99:1484314848 28. Schuster C, Gerold KD, Schober K, Probst L, Boerner K, Kim MJ, Ruckdeschel A, Serwold T, Kissler S: The autoimmunityassociated gene CLEC16A modulates thymic epithelial cell autophagy and alters T cell selection. Immunity 2015;42:942952 29. Kim S, Naylor SA, DiAntonio A: Drosophila Golgi membrane protein Ema promotes autophagosomal growth and function. Proceedings of the National Academy of Sciences 2012;109:E1072–E1081 30. Ardley HC, Robinson PA: E3 ubiquitin ligases. Essays Biochem 2005;41:1530 31. Birsa N, Norkett R, Wauer T, Mevissen TET, Wu HC, Foltynie T, Bhatia K, Hirst WD, Komander D, PlunFavreau H, Kittler JT: Lysine 27 Ubiquitination of the Mitochondrial Transport Protein Miro Is Dependent on Serine 65 of the Parkin Ubiquitin Ligase. Journal of Biological Chemistry 2014;289:1456914582 32. Fu M, StPierre P, Shankar J, Wang PT, Joshi B, Nabi IR: Regulation of mitophagy by the Gp78 E3 ubiquitin ligase. Molecular biology of the cell 2013;24:11531162 33. Mizuno E, Iura T, Mukai A, Yoshimori T, Kitamura N, Komada M: Regulation of Epidermal Growth Factor Receptor DownRegulation by UBPYmediated Deubiquitination at Endosomes. Mol Biol Cell 2005;16:51635174 34. Narendra D, Tanaka A, Suen DF, Youle RJ: Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. The Journal of Cell Biology 2008;183:795803

24 Page 25 of 46 Diabetes

35. Soderberg O, Gullberg M, Jarvius M, Ridderstrale K, Leuchowius KJ, Jarvius J, Wester K, Hydbring P, Bahram F, Larsson LG, Landegren U: Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat Meth 2006;3:9951000 36. Molina AJA, Wikstrom JD, Stiles L, Las G, Mohamed H, Elorza A, Walzer G, Twig G, Katz S, Corkey BE, Shirihai OS: Mitochondrial Networking Protects βCells From Nutrient Induced Apoptosis. Diabetes 2009;58:23032315 37. Poitout V, Robertson RP: Glucolipotoxicity: Fuel Excess and βCell Dysfunction. Endocr Rev 2008;29:351366 38. ShibaFukushima K, Imai Y, Yoshida S, Ishihama Y, Kanao T, Sato S, Hattori N: PINK1mediated phosphorylation of the Parkin ubiquitinlike domain primes mitochondrial translocation of Parkin and regulates mitophagy. Scientific Reports 2012;2:1002 39. Cornelissen T, Haddad D, Wauters F, Van Humbeeck C, Mandemakers W, Koentjoro B, Sue C, Gevaert K, De Strooper B, Verstreken P, Vandenberghe W: The deubiquitinase USP15 antagonizes Parkinmediated mitochondrial ubiquitination and mitophagy. Human Molecular Genetics 2014;23:52275242 40. Cunningham CN, Baughman JM, Phu L, Tea JS, Yu C, Coons M, Kirkpatrick DS, Bingol B, Corn JE: USP30 and parkin homeostatically regulate atypical ubiquitin chains on mitochondria. Nat Cell Biol 2015;17:160169 41. Bingol B, Tea JS, Phu L, Reichelt M, Bakalarski CE, Song Q, Foreman O, Kirkpatrick DS, Sheng M: The mitochondrial deubiquitinase USP30 opposes parkinmediated mitophagy. Nature 2014;510:370375 42. De Ceuninck L, Wauman J, Masschaele D, Peelman F, Tavernier J: Reciprocal cross regulation between RNF41 and USP8 controls cytokine receptor sorting and processing. Journal of Cell Science 2013;126:37703781 43. Finn RD, Bateman A, Clements J, Coggill P, Eberhardt RY, Eddy SR, Heger A, Hetherington K, Holm L, Mistry J, Sonnhammer ELL, Tate J, Punta M: Pfam: the protein families database. Nucleic Acids Res 2014;42:D222D230 44. Hatakeyama J, Wald JH, Rafidi H, Cuevas A, Sweeney C, Carraway KL: The ER structural protein Rtn4A stabilizes and enhances signaling through the receptor tyrosine kinase ErbB3. Science Signaling 2016;9:ra65ra65 45. Byun S, Shin SH, Lee E, Lee J, Lee SY, Farrand L, Jung SK, Cho YY, Um SJ, Sin H S, Kwon YJ, Zhang C, Tsang BK, Bode AM, Lee HJ, Lee KW, Dong Z: The retinoic acid derivative, ABPN, inhibits pancreatic cancer through induction of Nrdp1. Carcinogenesis 2015;36:15801589 46. Bonal CB, Baronnier DE, Pot C, Benkhoucha M, Schwab ME, Lalive PH, Herrera PL: NogoA Downregulation Improves Insulin Secretion in Mice. Diabetes 2013;62:14431452 47. Fujimaki T, Kato K, Yokoi K, Oguri M, Yoshida T, Watanabe S, Metoki N, Yoshida H, Satoh K, Aoyagi Y, Nozawa Y, Kimura G, Yamada Y: Association of genetic variants in SEMA3F, CLEC16A, LAMA3, and PCSK2 with myocardial infarction in Japanese individuals. Atherosclerosis 2010;210:468473 48. Berge T, Leikfoss I, Harbo H: From Identification to Characterization of the Susceptibility Gene CLEC16A. International Journal of Molecular Sciences 2013;14:4476 49. Redmann V, Lamb CA, Hwang S, Orchard RC, Kim S, Razi M, Milam A, Park S, Yokoyama CC, Kambal A, Kreamalmeyer D, Bosch MK, Xiao M, Green K, Kim J, Pruett Miller SM, Ornitz DM, Allen PM, Beatty WL, Schmidt RE, DiAntonio A, Tooze SA, Virgin HW: Clec16a is Critical for Autolysosome Function and Purkinje Cell Survival. Scientific Reports 2016;6:23326

25 Diabetes Page 26 of 46

50. Yang M, Chen T, Li X, Yu Z, Tang S, Wang C, Gu Y, Liu Y, Xu S, Li W, Zhang X, Wang J, Cao X: K33linked polyubiquitination of Zap70 by Nrdp1 controls CD8+ T cell activation. 2015;16:1253

26 Page 27 of 46 Diabetes

FIGURE LEGENDS

Figure 1. The selective ubiquitin ligase inhibitor lenalidomide impairs βcell function,

Nrdp1 ubiquitination, and mitophagy.

(A) Calculated HOMAβ measured in multiple myeloma patients pre and 12weeks post

initiation of chemotherapy (n=11/control, 14/lenalidomide). (B) Glucose stimulated insulin

secretion following static incubations in 1.67mM and 16.7mM glucose performed in isolated

CD1 islets pretreated with vehicle (DMSO) or 10 µM lenalidomide for 24hr (n=4/group).

(C) Fold oxygen consumption rates (OCR) in isolated islets pretreated with vehicle or 10

µM lenalidomide for 24hr prior to dispersion to single cells for analysis as adherent

monolayers on Seahorse flux analyzer (n=10/group). (D) Representative images of pancreas

sections from C57BL/6 mice treated for 3weeks in vivo with 50mg/kg lenalidomide or

vehicle alone, stained for insulin (cyan), LC3 (green) and SHDA (red). Nuclei are

demarcated with DAPI (blue). n=4/group (E) Quantification of LC3/SDHA colocalization

and total LC3+ puncta in islets from C57BL/6 mice treated in vivo with (black bars) or

without (white bars) 50mg/kg Lenalidomide (n=4/group, ~900 βcells and ~25,000 total

LC3/SDHA structures quantified/group). (F) Representative Western blot (WB) of LC3

levels in isolated islets treated with vehicle or 10 µM lenalidomide for 24hr with

quantification (by densitometry) of LC3II levels normalized to cyclophilin B. n=4/group.

(G) Representative deconvolution images of Min6 βcells (transfected with LC3GFP and

mitodsRed plasmids), then treated with vehicle or 10 µM lenalidomide for 24hr.with

addition of 5µM FCCP for the final 4hrs. n=4/group. (H) Quantification of LC3GFP/mito

dsRed and mitodsRed colocalization and total LC3GFP puncta in Min6 βcells treated with

vehicle or 10 µM lenalidomide for 24hr with addition of 5µM FCCP for the final 4hrs.

(n=4/group, ~1500 total GFP and dsRed puncta quantified per experiment). *p<0.05,

27 Diabetes Page 28 of 46

**p<0.01, ***p<0.001 by nonpaired student’s ttest, $$p<0.01 by paired student’s ttest, respectively.

2. Clec16a promotes the ubiquitination of Nrdp1.

(A) Representative WB of in vivo ubiquitination assay of overexpressed HAtagged Nrdp1 performed in HEK293T cells transfected with MycUbiquitin, Flag taggedempty vector

(EV), Clec16a or USP8 (as well as whole cell lysate HANrdp1, FlagClec16a, and Flag

USP8 levels) treated with DMSO () or 20 µM MG132 (+) overnight. n=3/group. (B)

Representative WB of in vivo ubiquitination assay of endogenous Nrdp1 performed in 293T cells transfected with MycUbiquitin and FlagEV or FlagClec16a (as well as whole cell lysate Nrdp1 and FlagClec16a levels). n=3/group. (C) Representative WB of in vivo endogenous ubiquitination assay of endogenous Nrdp1 ubiquitination performed in primary dermal fibroblasts derived from WT and Clec16a deficient mice (as well as whole cell lysate

Nrdp1 levels). n=3/group (D) Representative WB of in vivo ubiquitination assay of overexpressed HAtagged wildtype (WT) or C34S H36Q mutant (CSHQ) Nrdp1 performed in HEK293T cells transfected with MycUbiquitin and FlagEV or FlagClec16a (as well as whole cell lysate HANrdp1 and FlagClec16a levels). n=3/group. (E) Representative WB of in vivo ubiquitination assay of overexpressed HAtagged Nrdp1 performed in HEK293T cells transfected with MycUbiquitin and FlagEV or FlagClec16a (as well as whole cell lysate

HANrdp1 and FlagClec16a levels) treated with DMSO () or 10 µM lenalidomide (+) for

24 hr. n=3/group. (F) Glucose stimulated insulin secretion following static incubations in

1.67mM and 16.7mM glucose performed in isolated MIPCreERT2 control or βClec16aKO islets pretreated with vehicle (DMSO) or 10 µM lenalidomide for 24hr (n=4/group). *p<0.05 nonpaired student’s ttest.

28 Page 29 of 46 Diabetes

3. Clec16a possesses E3 ligase activity and directly ubiquitinates Nrdp1 via a nonK48

linkage.

(A) Representative αFlag WB from purified Clec16a6xHisFlag protein following

incubation in vitro with ATP, Ubiquitin, E1, and 26 unique E2conjugating enzymes at 37°C

for 1 hour. Arrows demarcate both the expected and novel high molecular weight Clec16a

proteins identified by WB. n=3/group. (BC) Representative WB of in vitro ubiquitination

assay of recombinant GSTClec16aFlag (B) or Clec16a6xHisFlag protein (C) following

incubation with ATP, HAUb, E1, in the presence or absence of E2 conjugation enzymes

UbE2D3 and/or UbE2c at 37°C for 1 hour. n=3/group. (D) Representative WB of in vitro

ubiquitination assay of recombinant GSTNrdp1CSHQ following incubation with ATP, HA

Ub, E1, in the presence or absence of UbE2D3 and recombinant Clec16a6xHisFlag at 37°C

for 1 hour. n=3/group. (E) Representative WB of in vivo ubiquitination assay of

overexpressed myctagged Nrdp1 performed in HEK293T cells transfected with FlagEV or

FlagClec16a in the presence of HAtagged wildtype (WT) ubiquitin as well as ubiquitin

mutants with all lysines converted to arginines (KO), ubiquitin only capable of K48linked

ubiquitination (K48), ubiquitin incapable of K48linked ubiquitination (K48R), or ubiquitin

only capable of K63linked ubiquitination (K63) (as well as whole cell lysate mycNrdp1 and

FlagClec16a levels). n=3/group.

4. Ubiquitination is necessary to maintain the Clec16aNrdp1USP8 protein complex.

(A) Representative WB following cell fractionation and centrifugation of stably expressing

FlagClec16a NIH3T3 cells probed for Flag (Clec16a), endogenous Nrdp1, endogenous

USP8 as well as porin (mitochondria), Creb (nuclei), and Rab5 (endosomal/soluble small

GTPase) as markers of cell compartments. n=3/group. (B) Representative deconvolution

image of NIH3T3 fibroblasts following transfection with GFPClec16a, FlagUSP8, and HA

29 Diabetes Page 30 of 46

Nrdp1 plasmids with indirect immunofluorescence staining for GFPClec16a (green), Flag

USP8 (red), and HANrdp1 (blue). Inset – triple colocalized structures (white) highlighted with yellow arrows. n=3/group. (C) Representative WB of reticulocyte lysates following in vitro transcription/translation of FlagClec16a and HANrdp1 with antiHA (left) or antiFlag

(right) IP. n=3/group. (D) Representative WB of reticulocyte lysates, following in vitro transcription/translation of FlagClec16a and HANrdp1 protein treated in solution with 75

µM PYR41 at 37°C for 3 hours, following antiFlag IP. n=3/group. (E) Representative WB following antimyc IP in HEK293T cells transfected with MycNrdp1, FlagUSP8 and either

GFPEV or –Clec16a, along with WT or KO Ubiquitin vectors. n=3/group. (F)

Representative WB of in vivo K6linkage specific ubiquitination assay of overexpressed

Parkin performed in HEK293T cells transfected with HAK6Ubiquitin, FlagEV or USP8, and GFPEV orClec16a (as well as whole cell lysate GFPNrdp1, FlagUSP8, and Parkin levels). n=3/group.

5. Specific Nrdp1 ubiquitination is vital for βcell function.

(A) Representative WB of in vivo ubiquitination assay of overexpressed HAtagged wildtype

(WT) or panK to R mutant (KR) Nrdp1 performed in HEK293T cells transfected with Myc

Ubiquitin and FlagEV or FlagClec16a (as well as whole cell lysate HANrdp1 and Flag

Clec16a levels). n=3/group. (B) Representative WB of endogenous USP8 following anti

FlagClec16a IP in HeLa cells transfected with FlagEV or FlagClec16a and HAEV or HA

Nrdp1 expression vectors (WT, CSHQ, or KR). n=3/group. (C) Representative WB of HA

Nrdp1 levels in Min6 βcells, 72 hours after transfection with nontargeting (NT) or Clec16a

specific shRNA plasmids, as well as HAEV, HANrdp1 WT, or HANrdp1 KR.

(n=5/group). (D) Glucose stimulated insulin secretion following static incubations in 2 mM and 20 mM glucose performed in Min6 βcells, 72 hours after transfection with non

30 Page 31 of 46 Diabetes

targeting (NT) or Clec16a specific shRNA plasmids, as well as HAEV, HANrdp1 WT, or

HANrdp1 KR. (n=5/group). (E) Top – Representative deconvolution image of Nrdp1:USP8

proximity ligation assay (PLA – red, DAPI blue) in Min6 βcells, 24 hours after treatment

with vehicle (DMSO) or 10 μ M lenalidomide. Bottom quantification of relative

Nrdp1:USP8 PLA events in Min6 βcells (fold change vs. vehicle controls) following

treatment conditions as noted. (n=3/group; ~2900 PLA events quantified/group per

experiment).

6. The Clec16aNrdp1USP8 complex is perturbed by glucolipotoxicity.

(A) Top – Representative deconvolution image of Nrdp1:USP8 proximity ligation assay

(PLA – red, DAPI blue) in Min6 βcells, 72 hours after transfection with nontargeting

(NT) or Clec16a specific shRNA plasmids, following treatment with BSA control or 0.4 mM

palmitate for 48 hr. Bottom quantification of relative Nrdp1:USP8 PLA events in Min6 β

cells (fold change vs. shNT BSA controls) following treatment conditions as noted.

(n=4/group; ~1200 PLA events quantified/group per experiment). (B) Top – Representative

deconvolution image of Nrdp1:USP8 proximity ligation assay (PLA – red, PDX1 – green,

DAPI blue) in βcells of dissociated human islets following 48 hr treatment with BSA

control (BSA) or 0.4mM palmitate/0.92% BSA + 20mM glucose (palmitate). Bottom –

quantification of relative Nrdp1:USP8 PLA events in βcells of dissociated human islets

after control BSA (white bar) or 0.4mM palmitate/0.92% BSA + 20mM glucose (grey bar)

treatment for 48 hr. (n=3 donors; ~900 βcell PLA events quantified/group per donor). (C)

Top – Representative WB of Nrdp1 levels in isolated mouse islets treated for 48 hr with or

without 0.4mM palmitate+20mM glucose. Bottom relative quantification (by densitometry)

of Nrdp1 levels by WB in isolated islets from mice treated for 48 hr with (grey bars) or

31 Diabetes Page 32 of 46

without (white bars) 0.4mM palmitate/0.92% BSA + 20mM glucose. n=3/group. (D)

Representative WB of in vivo ubiquitination assay of HAtagged Nrdp1 in MIN6 cells transfected with HANrdp1, MycUb and FlagEV or FlagClec16a plasmids, and treated for

48 hr with or without 0.4mM palmitate/BSA coupling. n=3/group (E) Representative WB following FlagIP in MIN6 cells transfected with HANrdp1, FlagUSP8, and either GFPEV or GFPClec16a plasmids and treated for 48 hr with or without 0.4mM palmitate. n=4/group.

(F) Top – Representative WB of Cleaved Caspase3 levels in MIN6 cells transfected with or without GFPClec16a, and 48hr BSA or Palmitate treatment. Bottom – relative quantification

(by densitometry) of Cleaved Caspase3 levels (normalized to Actin loading control) by WB in MIN6 cells transfected with or without GFPClec16a and treated for 48hr with BSA (white bars) or Palmitate (grey bars) couplings. n=5/group. #p<0.05 paired student’s ttest. *p<0.05,

**p<0.01 ***p<0.001 nonpaired student’s ttest.

7. Schematic model of the Clec16aNrdp1USP8 mitophagy complex.

(A) Under physiological conditions, Clec16a promotes nondegradative ubiquitination of the

E3ligase Nrdp1 to facilitate complex formation between Clec16aUSP8Nrdp1. This complex functions to promote normal mitophagy initiation and balance mitochondrial quality control, which in turn leads to normal βcell function and regulated insulin release. (B)

However, under conditions of impaired Clec16a function, such as pharmacologic inhibition by the chemotherapeutic lenalidomide or glucolipotoxicity, Clec16amediated Nrdp1 ubiquitination is inhibited and the Clec16aUSP8Nrdp1 complex is destabilized. This leads to dysfunctional mitophagy, loss of mitophagic quality control, and dysregulated mitochondrial respiration, ultimately leading to compromised βcell insulin release.

32 FIGUREPage 33 of1 46 Diabetes A. B. Vehicle C. Vehicle Control Lenalidomide Lenalidomide Lenalidomide §§ * ** *** Fold OCR HOMA-ß (%)

1.67mM16.7mM 5μM PRE POST PRE POST 1.67mM 16.7mM glucose glucose FCCP

Insulin secretion (pg/ng insulin content/hr) glucose glucose D. E. Vehicle Lenalidomide INSULIN LC3 SDHA MERGE * Vehicle

Co-localization (%LC3/SDHA) * Lenalidomide LC3 puncta/cell

F. G. H. Vehicle Lenalidomide Vehicle FCCP Vehicle Lenalidomide Vehicle Lenalidomide Lenalidomide - + - + * LC3 CyclophilinB GFP-LC3 GFP-LC3 GFP-LC3 GFP-LC3 Vehicle Lenalidomide Vehicle FCCP Co-localization (%LC3-GFP/mito-dsRed)

mito-dsRed mito-dsRed mito-dsRed mito-dsRed * LC3-II (Fold change Vehicle) LC3-GFP LC3-GFP puncta/cell

MERGE MERGE MERGE MERGE Vehicle FCCP FIGURE 2 Diabetes Page 34 of 46 A.A. B. C. KO MG132 - +-- + +

WT Clec16a IP: HA-Nrdp1 IP: Myc-Ub WB: Myc-Ub WB: Nrdp1 IP: Nrdp1 WB: Ub

WB: Flag WB:Flag-Clec16a WB:HA-Nrdp1 WB:Nrdp1 INPUT

INPUT WB:Nrdp1 WB:Cyclophilin B WB:Cyclophilin B WB:Cyclophilin B Flag-Clec16a - + - - + - Flag-Clec16a - + INPUT Flag-USP8 -- + - - +

D. E. F. MIP-CreERT2 Vehicle Lenalidomide - +- + MIP-CreERT2 Lenalidomide ß-Clec16aKO Vehicle IP: Myc-Ub ß-Clec16aKO Lenalidomide WB: HA-Nrdp1 IP: HA-Nrdp1 WB: Myc-Ub * WB:HA-Nrdp1 WB:Flag-Clec16a

INPUT WB:Cyclophilin B WB:Flag-Clec16a Flag-Clec16a - + - + WB:HA-Nrdp1 HA-Nrdp1 WT + + -- HA-Nrdp1 CSHQ -- + + INPUT WB:CyclophilinB Insulin secretion (pg/ng insulin content/hr) 1.67mM 16.7mM glucose glucose FIGURE 3 A.Page 35 of 46 Diabetes B. E2 enzyme: A B C D1 D2 D3 D4 E1E2 WB: 170kDa GST-Clec16a-Flag + + + Clec16a-His-Flag 130kDa HA-Ub - + + UbE2D3 - ++ E2 enzyme: F G1 H J2 K L3 170kDa WB: 2J1 2V/2V12V/2V2 170kDa IP:Flag-Clec16a Clec16a-His-Flag 130kDa 130kDa WB: HA-Ub

E2 enzyme: Z W2 T S R2 R1 Q2-1Q1-2 WB: 170kDa WB: Flag-Clec16a

Clec16a-His-Flag 130kDa INPUT

C. UbE2C UbE2D3 D. Clec16a-His-Flag + + + + + + Clec16a-His-Flag - + - + ATP + + - + + - UbE2D3 -- + + E2 + - + + - + GST-Nrdp1 CSHQ + + + +

100kDa IP:Nrdp1 75kDa IP: Flag-Clec16a WB: HA-Ub WB: HA-Ub 170kDa 50kDa 130kDa 37kDa 100kDa WB: Flag-Clec16a 70kDa

INPUT WB: HA-Nrdp1 WB: Flag-Clec16a INPUT E.

Ubiquitin: WT WT KO K48 K48R K63

IP: Myc-Nrdp1 WB: HA-Ub

WB:Flag-Clec16a

WB: Myc-Nrdp1 INPUT WB:Cyclophilin B Flag-Clec16a - + + + + + FIGURE 4 Diabetes Page 36 of 46 A. B. C.

600g 8000g 100000g WC S P S P S

WB:Flag-Clec16a Flag-USP8 WB:USP8 GFP-Clec16a IP:IgG IP:IgG IP:Flag INPUT IP:HA INPUT WB:Nrdp1 WB:Flag-Clec16a WB:Porin WB: HA-Nrdp1 WB:Creb Merge WB:Rab5 HA-Nrdp1

D. E. F.

Ubiquitin WT WT KO KO KO PYR41 - + IP: Myc-Nrdp1 WB: HA-Nrdp1 WB: Flag-USP8 IP:Parkin WB: Flag-Clec16a WB: HA-K6-Ub IP:Flag IP: Myc-Nrdp1 WB: GFP-Clec16a WB:HA-Nrdp1 WB:Flag-Clec16a WB:Flag-USP8 WB:GFP-Clec16a WB:GFP-Clec16a WB: Flag-USP8 INPUT Ponceau Red INPUT WB:Myc-Nrdp1 WB:Parkin INPUT WB:Cyclophilin B WB:Cyclophilin B GFP-Clec16a - + - + ++ GFP-Clec16a - +- + Flag-USP8 - + - + FIGUREPage 37 of 5 46 Diabetes A. B. C. IP: Flag-Clec16a WB: USP8 WB: HA-Nrdp1 IP: HA-Nrdp1 WB:Cyclophillin B WB: Myc-Ub WB: USP8 NT shRNA +-++ - - WB:Flag-Clec16a Clec16a shRNA - - - + + + HA-Nrdp1 WT WB: HA-Nrdp1 - -+ - -+ WB: HA-Nrdp1 INPUT HA-Nrdp1 K-R - - + - - + WB: CyclophilinB WB: Flag-Clec16a

INPUT Flag-Clec16a - + + + + WB: Cyclophilin B HA-Nrdp1 WT - - -+ - Flag-Clec16a - +- + HA-Nrdp1 K-R - - - + - HA-Nrdp1 WT + -+ - HA-Nrdp1 CSHQ - --- + HA-Nrdp1 K-R -- + + D. E. Vehicle Lenalidomide

2mM glucose # 20mM glucose # #

DAPI Nrdp1:USP8 PLA Vehicle Lenalidomide Insulin Secretion (Fold change 2mM glucose) ** NT shRNA -+- -++ + + + - -- Clec16a shRNA --- + + + --- + + + HA-Nrdp1 WT - -+- -+ - -+- -+

HA-Nrdp1 K-R --+--+ --+--+ Fold PLA Nrdp1:USP8 FIGURE 6 Diabetes Page 38 of 46 A. B. PDX-1/ BSA Palmitate Nrdp1:USP8 PLA PLA PLA DAPI DAPI NT BSA shRNA

Clec16a Palmitate shRNA PDX-1 *** Nrdp1:USP8 PLA * BSA Palmitate * BSA Palmitate Fold PLA Fold PLA Nrdp1:USP8 Fold PLA Fold PLA Nrdp1:USP8 NT Clec16a shRNA shRNA C. D. Palmitate - + - + Palmitate - +- + Nrdp-1 IP: Myc-Ub Cyclophilin B WB: HA-Nrdp1

BSA Palmitate WB:HA-Nrdp1 WB:Flag-Clec16a INPUT *** WB:Cyclophilin B Flag-Clec16a - + - + Nrdp1 (Fold change BSA)

E. F. Palmitate - - + + Cleaved Caspase-3 BSA Palmitate GFP-Clec16a IP: Flag-USP8 WB: HA-Nrdp1 Actin

WB:HA-Nrdp1 *** * WB:GFP-Clec16a

INPUT WB:Flag-USP8 ns WB:Cyclophilin B

GFP-Clec16a - + - + Cleaved Caspase-3 (Fold change EV-BSA) GFP-Clec16a - + - + FIGUREPage 39 of7 46 Diabetes A. Degradative Ub B. Non-degradative Ub

Clec16a Clec16a Lenalidomide Nrdp1 Palmitate USP8 Nrdp1 USP8

Regulated mitophagy: Abberant mitophagy: Healthy mitochondria Accumulation of unhealthy mitochondria

Normal ß-cell function Impaired ß-cell function Diabetes Page 40 of 46

Supplementary Table 1. List of antibodies.

Antibody Company Catalog number/clone number (if applicable) CREB Cell Signaling Technology 9197 (clone 48H2) Cyclophilin B Pierce PA1027 FLAG SigmaAldrich F1804 (clone M2) GFP Abcam ab6673 HA Santa Cruz Biotechnology sc805 (clone Y11) HAHRP Roche 12 013 819 001 (clone 3F10) Insulin Abcam ab7842 LC3 SigmaAldrich L8918 Myc Developmental Studies clone 9E10 Hybridoma Bank Nrdp1 Santa Cruz Biotechnology sc79996 (clone E14) Nrdp1 Bethyl Laboratories Inc A300049A (FLRF/RNF41) Parkin EMD Millipore 05882 (clone PRK8) Pdx1 Santa Cruz sc14664 (clone A17) Rab5 Cell Signaling Technology 3547 (clone C8B1) SDHA Abcam ab14715 (clone 2E3GC12FB2AE2) Ubiquitin Enzo Life Sciences BMLPW88100500 (clone FK2) USP8 SigmaAldrich SAB4200527 (clone US872)

Supplementary Table 2. Donor Demographics for human islets.

Donor Age Sex Ethnicity BMI Cause of Death #1 32 Female Latino 27.9 Anoxia #2 54 Male White 35.6 Cerebrovascular/stroke #3 56 Female Latino 22.7 Cerebrovascular/stroke

Supplementary Table 3. Patient demographics for control or Lenalidomide treatment groups.

CONTROL LENALIDOMIDE pvalue n 11 14 NA Age (yrs) 66.3±2.1 55.9±3.6 * 0.021 Gender 7M 4F 10M 4F NA Weight (kg) 91.3±6.9 83.3±6.1 0.399

Page 41 of 46 Diabetes

Supplementary Figure 1. Lenalidomide treatment in mice in vivo impairs glucose

tolerance. (a) Blood glucose concentrations during IPGTT of C57B6 mice treated for 3

weeks with 50mg/kg I.P. Lenalidomide (white circles) or vehicle control (black circles). (n=7

8/group) (b) Blood glucose area under the curve (AUC) from IPGTT of C57B6 mice treated

for 3weeks with 50mg/kg I.P. Lenalidomide (white bar) or vehicle control (black bar). (n=7

8/group). (c) Representative Western blot of in vivo endogenous ubiquitination assay of

endogenous Nrdp1 (with whole cell lysate Nrdp1 levels) in Min6 βcells treated with vehicle

or 10M lenalidomide for 24hr. Cyclophilin B serves as a loading control. (n=3/group). ##

p<0.01 twoway ANOVA, with Sidak’s multiple comparison posttest. * p<0.05 nonpaired

student’s ttest. Diabetes Page 42 of 46

SUPPLEMENTARY FIGURE 2 A. B. C. flox NT lec16aNA WT Clec16a C iR siRNA s Nrdp1 Cyclophilin B CHX (hr) 02 4 6 024 6

** ***

** Fold Clec16a/HPRT mRNA flox WT Clec16a WT flox NT Clec16a Clec16a FoldClec16a/HPRT mRNA siRNA siRNA Nrdp1expression (% baseline) Time (hr)

D. MG132 + +++ E. F. Nrdp1 Nrdp1 DMSO CQ BA Clec16a Nrdp1 Clec16a USP8 Clec16a USP8 Cyclophilin B Cyclophilin B FlagClec16a + + + FlagClec16a + + + Cyclophilin B shUSP81 + + GFPClec16a + + + + shUSP82 + + FlagUSP8 + + + +

Supplementary Figure 2. Clec16a stabilizes Nrdp1 independent of USP8 or lysosomal function. (a) Endogenous Nrdp1 levels by representative WB (top) and Clec16a mRNA levels by qRTPCR (bottom) measured in Min6 βcells, 72 hours after transfection with non targeting (NT) or Clec16a specific siRNA. (b) Clec16a mRNA levels by qRTPCR from RNA isolated from WT or UbEcreERT2;Clec16aflox/flox KO primary dermal fibroblasts. n=3/group.

(c) Representative WB of endogenous Nrdp1 levels (with densitometry of % change from basal levels) from WT or Clec16aflox KO primary dermal fibroblasts following treatment with cycloheximide (CHX) for 06 hrs. n=3/group. (d) Representative WB of HANrdp1, GFP

Clec16a and FlagUSP8 levels in HEK293T cells, 48 hours after transfection with GFPEV or

GFPClec16a and FlagEV or FlagUSP8, following overnight treatment with vehicle (DMSO) or 20M MG132. n=3/group. (e) Representative WB of HANrdp1, FlagClec16a and endogenous USP8 levels in HeLa cells, 48 hours after transfection with FlagEV or Flag

Clec16a and/or 2 independent USP8 shRNA vectors (or NT controls). n=3/group. (f)

Representative WB of HANrdp1 and FlagClec16a protein levels in HEK293T cells, 48 hours after transfection with FlagEV or FlagClec16a, following treatment with vehicle Page 43 of 46 Diabetes

(DMSO), chloroquine (CQ; 10M) or Bafilomycin A1 (BA; 150nM) for 6 hours. n=3/group.

*p<0.05 twoway ANOVA with Sidak’s multiple comparison posttest. **p<0.01 nonpaired

student’s ttest.

Diabetes Page 44 of 46

Supplementary Figure 3. Inhibition of Clec16a function in βcells by pharmacologic or genetic approaches. (a) Clec16a mRNA levels by qRTPCR from RNA isolated from 2 monthold MIPCreERT2 or βClec16aKO mice. (n=4/group). (b) Blood glucose concentrations during IPGTT of 5monthold MIPCreERT2 or βClec16aKO mice.

(n=4/group) p<0.01 twoway ANOVA. (c) Blood glucose area under the curve (AUC) from

IPGTT of 5monthold MIPCreERT2 or βClec16aKO mice. (n=4/group) (d) Clec16a mRNA levels by qRTPCR from RNA isolated from isolated islets treated for 24hr with vehicle

(DMSO) or 10M Lenalidomide. (n=4/group). (e) Clec16a mRNA levels by qRTPCR from

RNA isolated from Min6 cells, 72 hours after transfection with nontargeting (NT) or one of two independent Clec16aspecific shRNA (Clec16a1, Clec16a2). (n=4/group). (f) Glucose stimulated insulin secretion following static incubations in 2.5 mM and 25 mM glucose performed in Min6 βcells, 72 hours after transfection with nontargeting or Clec16aspecific shRNA plasmids. (n=4/group). ##p<0.01, ###p<0.001 twoway ANOVA, Tukey’s multiple comparison posttest. ***p<0.001 oneway ANOVA, Tukey’s multiple comparison post test.**p<0.01 nonpaired student’s ttest.

Page 45 of 46 Diabetes

Plasmids

Plasmids used for transfections included pFLAGCMV5aClec16a, pcDNA3.1 3xHANrdp1,

MycUbiquitin, pBABEpuroLC3GFP (Addgene plasmid 22405, from the lab of Dr. Jayanta

Debnath), mCherryParkin (Addgene plasmid 23956, from the lab of Dr. Richard Youle), and

pDsRed2mito (Clontech), all of which were previously described (1). pFLAGCMV5a

Clec16a6xHis was generated by insertion of annealed oligos bearing the sequences (5’

TCGACCACCACCACCACCACCACGGTAC3’ and 5’ CGTGGTGGTGGTGGTGGTGG 3’)

into a SalIKpnI restriction site of the pFLAGCMV5aClec16a vector. Clec16aEGFP was

generated by ligation of the Clec16a ORF insert (derived from the pFLAGCMV5aClec16a

vector) into the mEGFPN1 vector (Addgene #54767, a gift from Dr. Michael Davidson).

pGSTagClec16aFlag and pGSTagNrdp1 were generated by ligation of the Clec16a ORF

Flag fusion protein or the Nrdp1 cDNA into the pGSTag vector (Addgene #21877, a gift from

Dr. David Ron). pcDNA3.1MycNrdp1 was generated by insertion of annealed oligos

bearing the sequences (5’

AGCTTGCCACCATGGAACAAAAACTCATCTCAGAAGAGGATCTGG3’ and 5’

GATCCCAGATCCTCTTCTGAGATGAGTTTTTGTTCCATGGTGGCA3’) into a HindIII

BamHI restriction site of the pcDNA3.13xHANrdp1 vector, thereby eliminating the 3xHA

tag sequence. pcDNA3.13xHANrdp1 C34S H36Q (CSHQ) was generated by sitedirected

mutagenesis of DNA sequences encoding the amino acids of interest utilizing the

QuikChange II Sitedirected mutagenesis kit per the manufacturer’s protocols (Agilent).

Primers used for Nrdp1 CSHQ sitedirected mutagenesis included 5’

TTGCAGAAGGCCTGCTCACTATGAGGCGCCTGCAC3’ and 5’

GTGCAGGCGCCTCATAGTGAGCAGGCCTTCTGCAA3’. The pcDNA3.13xHANrdp1 K

R vector (pan lysine to arginine mutant) was generated by gene synthesis approaches to

generate the 3xHANrdp1 sequence bearing modification DNA encoding all 11 lysine

residues to mutated to arginine residues (Life Technologies). This insert was subsequently

subcloned into the pcDNA3.1 vector (Life Technologies). The full sequence of this gene Diabetes Page 46 of 46

synthesis insert is available upon request. pcDNADest40Clec16aFlag was generated by

Gateway cloning recombination of the Clec16aFlag insert from the previously described pBABEpuroDestClec16aFlag vector (1) into the pcDNADest40 destination vector (Life

Technologies) using the manufacturer’s protocols. An expression vector for USP8 was generated by PCR amplification of cDNA isolated from Min6 mouse insulinoma cells. PCR primers to amplify USP8 included restriction enzyme sites to subclone coding sequences in expression plasmids. USP8 was subcloned into pFLAGCMV5a (Sigma). Primer sequences were as follows, 5’AGATCTGCCACCATGCCTGCTGTAGCTTCAGTTCCTAAA3’ and 5’

GTCGACTGTGGCTACATCAGTTATGCGTGGTCCCAGGGA3’. Additional plasmids included pRK5HAUbiquitinWT, pRK5HAUbiquitinKO, pRK5HAUbiquitinK48R, pRK5

HAUbiquitinK48, and pRK5HAUbiquitinK63 (Addgene plasmids 1760317608, a gift from the lab of Dr. Ted Dawson) as well as pRK5HAUbiquitinK6 (Addgene plasmids 22900, a gift from the lab of Dr. Sandra Weller). USP8 shRNA sequences included:

5’CCGGTCAAGCAACAGCAGGATTATTCTCGAGAATAATCCTGCTGTTGCTTGATTTTTG

3’

5’CCGGGCTGTGTTACTAGCACTATATCTCGAGATATAGTGCTAGTAACACAGCTTTTTG

3’. Nontargeting and Clec16a siRNA and shRNA sequences were previously published (1).

Plasmid sequences were confirmed by DNA sequencing.

REFERENCES

1. Soleimanpour Scott A, Gupta A, Bakay M, Ferrari Alana M, Groff David N, Fadista J, Spruce Lynn A, Kushner Jake A, Groop L, Seeholzer Steven H, Kaufman Brett A, Hakonarson H, Stoffers Doris A: The Diabetes Susceptibility Gene Clec16a Regulates Mitophagy. Cell 2014;157:15771590